Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.023
Filter
1.
J Cell Mol Med ; 28(19): e70143, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39383106

ABSTRACT

Sad and UNC84 domain 1 (SUN1) is a kind of nuclear envelope protein with established involvement in cellular processes, including nuclear motility and meiosis. SUN1 plays an intriguing role in human adipose-derived stem cells (hASCs) differentiation; however, this role remains largely undefined. This study was undertaken to investigate the role of SUN1 in hASCs differentiation, as well as its underlying mechanisms. Employing siRNAs, we selectively downregulated SUN1 and CD36 expression. Microtubules were depolymerized using nocodazole, and PPARγ was activated using rosiglitazone. Western blotting was performed to quantify SUN1, PPARγ, α-tubulin, CD36, OPN, and adiponectin protein expression levels. Alkaline phosphatase and Oil red O staining were used to assess osteogenesis and adipogenesis, respectively. Downregulated SUN1 expression increased osteogenesis and decreased adipogenesis in hASCs, concomitant with upregulated α-tubulin expression and downregulated CD36 expression, alongside reduced nuclear localization of PPARγ. Microtubule depolymerization increased CD36 expression. Rescue experiments indicated that microtubule depolymerization counteracted the downregulated SUN1-induced phenotypic changes. This study demonstrates that SUN1 influences the differentiation of hASCs towards osteogenic and adipogenic lineages, indicating its essential role in cell fate.


Subject(s)
Adipogenesis , Adipose Tissue , CD36 Antigens , Cell Differentiation , Osteogenesis , PPAR gamma , Stem Cells , Tubulin , Humans , Adipogenesis/genetics , CD36 Antigens/metabolism , CD36 Antigens/genetics , Osteogenesis/genetics , Tubulin/metabolism , Stem Cells/metabolism , Stem Cells/cytology , PPAR gamma/metabolism , PPAR gamma/genetics , Adipose Tissue/cytology , Adipose Tissue/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Gene Expression Regulation , Cells, Cultured , Nuclear Proteins
2.
Sci Rep ; 14(1): 22988, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39362925

ABSTRACT

Tamoxifen (TAM) resistance is a major challenge in treating oestrogen receptor-positive (ER+) breast cancers. It is possible that the H2S synthase cystathionine-γ-lyase (CSE), which has been previously shown to promote tumour growth and metastasis in other cancer cells, is involved in this resistance. Therefore, we investigated CSE's role and potential mechanisms in TAM-resistant breast cancer cells. First, we examined the effect of CSE expression on TAM sensitivity and resistance in MCF7 (breast cancer) cells. The findings revealed that CSE was directly associated with TAM sensitivity and involved in TAM resistance in ER+ breast cancer cells, indicating that it may be useful as a biomarker. Next, we wanted to determine the molecular mechanism of CSE's role in TAM resistance. Using cell migration, co-immunoprecipitation, western blotting, and cell viability assays, we determined that the CSE/H2S system can affect the expression of PPARγ by promoting the sulfhydrylation of PPARγ, which regulates the transcriptional activity of ACSL1. ACSL1, in turn, influences STAT3 activation by affecting the phosphorylation, palmitoylation and dimerization of STAT3, ultimately leading to the development of TAM resistance in breast cancer. Finally, we examined the effect of CSE inhibitors on reducing drug resistance to determine whether CSE may be used as a biomarker of TAM resistance. We observed that the novel CSE inhibitor I194496 can reverse TAM resistance in TAM-resistant breast cancer via targeting the PPARγ/ACSL1/STAT3 signalling pathway. Overall, our data indicate that CSE may serve as a biomarker of TAM resistance and that the CSE inhibitor I194496 is a promising candidate for combating TAM resistance.


Subject(s)
Breast Neoplasms , Cystathionine gamma-Lyase , Drug Resistance, Neoplasm , PPAR gamma , Receptors, Estrogen , STAT3 Transcription Factor , Signal Transduction , Tamoxifen , Humans , Tamoxifen/pharmacology , Cystathionine gamma-Lyase/metabolism , STAT3 Transcription Factor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , PPAR gamma/metabolism , Female , Signal Transduction/drug effects , Receptors, Estrogen/metabolism , MCF-7 Cells , Cell Line, Tumor , Antineoplastic Agents, Hormonal/pharmacology , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects
3.
J Orthop Surg Res ; 19(1): 645, 2024 Oct 12.
Article in English | MEDLINE | ID: mdl-39396027

ABSTRACT

Our previous research revealed a close association between the acetylation of peroxisome proliferator-activated receptor γ (PPARγ) histone H3K27 and the adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). We preliminarily explored the epigenetic mechanism of steroid-induced avascular necrosis of the femoral head (SANFH) development, but the specific histone deacetylase (HDAC) involved in this regulatory process remains unknown. In this study, we combined cell, animal, and clinical specimen experiments to screen for specific HDAC genes that could regulate BMSC adipogenic differentiation and to explore their roles. The results showed that dexamethasone (DEX) significantly exacerbated the imbalance between the adipogenic and osteogenic differentiation of BMSCs, and there were differences in HDAC expression in the adipogenic differentiation cell models, with histone deacetylase 10 (HDAC10) showing the most significant decrease in expression. Subsequent use of a chromatin immunoprecipitation assay kit and quantitative polymerase chain reaction (ChIP‒qPCR) revealed a decrease in HDAC10 expression at predicted potential sites within the PPARγ promoter, indicating a significant decrease in HDAC10 enrichment in the PPARγ promoter region of BMSCs, thereby promoting sustained PPARγ expression. Additionally, immunohistochemistry of samples collected from mice and humans with SANFH and normal femoral heads revealed an imbalance between adipogenic and osteogenic differentiation in the necrotic area of femoral heads, with a significant decrease in the relative expression of HDAC10 in the necrotic area of femoral heads with SANFH. In summary, we speculate that HDAC10 affects the progression of SANFH by regulating BMSC adipogenic differentiation, a process possibly related to PPARγ histone acetylation. These findings provide a promising direction for the treatment of SANFH.


Subject(s)
Adipogenesis , Cell Differentiation , Dexamethasone , Femur Head Necrosis , Histone Deacetylases , Mesenchymal Stem Cells , PPAR gamma , Femur Head Necrosis/chemically induced , Femur Head Necrosis/pathology , Femur Head Necrosis/metabolism , Femur Head Necrosis/genetics , Animals , Mesenchymal Stem Cells/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Adipogenesis/drug effects , Adipogenesis/genetics , Cell Differentiation/drug effects , Humans , PPAR gamma/metabolism , PPAR gamma/genetics , Mice , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Male , Osteogenesis/drug effects , Osteogenesis/physiology , Osteogenesis/genetics , Cells, Cultured , Female
4.
Front Endocrinol (Lausanne) ; 15: 1394102, 2024.
Article in English | MEDLINE | ID: mdl-39398333

ABSTRACT

The PPARG gene encodes a member of a nuclear receptor superfamily known as peroxisome proliferator-activated gamma (PPARγ). PPARγ plays an essential role in adipogenesis, stimulating the differentiation of preadipocytes into adipocytes. Loss-of-function pathogenic variants in PPARG reduce the activity of the PPARγ receptor and can lead to severe metabolic consequences associated with familial partial lipodystrophy type 3 (FPLD3). This review focuses on recent scientific data related to FPLD3, including the role of PPARγ in adipose tissue metabolism and the phenotypic and clinical consequences of loss-of-function variants in the PPARG gene. The clinical features of 41 PPARG pathogenic variants associated with FPLD3 patients were reviewed, highlighting the genetic and clinical heterogeneity observed among 91 patients. Most of them were female, and the average age at the onset and diagnosis of lipoatrophy was 21 years and 33 years, respectively. Considering the metabolic profile, hypertriglyceridemia (91.9% of cases), diabetes (77%), hypertension (59.5%), polycystic ovary syndrome (58.2% of women), and metabolic-dysfunction-associated fatty liver disease (87,5%). We also discuss the current treatment for FPLD3. This review provides new data concerning the genetic and clinical heterogeneity in FPLD3 and highlights the importance of further understanding the genetics of this rare disease.


Subject(s)
Lipodystrophy, Familial Partial , PPAR gamma , Phenotype , Humans , Lipodystrophy, Familial Partial/genetics , Lipodystrophy, Familial Partial/pathology , PPAR gamma/genetics , Female , Loss of Function Mutation , Adipose Tissue/metabolism , Adipose Tissue/pathology
5.
Cells ; 13(19)2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39404417

ABSTRACT

The ongoing obesity epidemic has raised awareness of the complex physiology of adipose tissue. Abnormal adipocyte differentiation results in the development of systemic metabolic disorders such as insulin resistance and diabetes. The conjugation of NEDD8 (neural precursor cell expressed, developmentally downregulated 8) to target protein, termed neddylation, has been shown to mediate adipogenesis. However, much remains unknown about its role in adipogenesis. Here, we demonstrated that neddylation and its targets, the cullin (CUL) family members, are differentially regulated during mouse and human adipogenesis. Inhibition of neddylation by MLN4924 significantly reduced adipogenesis of 3T3-L1 and human stromal vascular cells. Deletion of NAE1, a subunit of the only NEDD8 E1 enzyme, suppressed neddylation and impaired adipogenesis. Neddylation deficiency did not affect mitotic cell expansion. Instead, it disrupted CREB/CEBPß/PPARγ signaling, essential for adipogenesis. Interestingly, among the neddylation-targeted CUL family members, deletion of CUL3, but not CUL1, CUL2, or CUL4A, largely replicated the adipogenic defects observed with neddylation deficiency. A PPARγ agonist minimally rescued the adipogenic defects caused by the deletion of NAE1 and CUL3. In conclusion, our study demonstrates that neddylation and its targeted CUL3 are crucial for adipogenesis. These findings provide potential targets for therapeutic intervention in obesity and metabolic disorders.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Cullin Proteins , NEDD8 Protein , Cullin Proteins/metabolism , Adipocytes/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Animals , Humans , Mice , Adipogenesis/drug effects , Adipogenesis/genetics , NEDD8 Protein/metabolism , NEDD8 Protein/genetics , Cell Differentiation/drug effects , Signal Transduction/drug effects , PPAR gamma/metabolism , Cyclopentanes/pharmacology , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes
6.
J Vis Exp ; (211)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39373467

ABSTRACT

Increasing levels of compounds have been detected in the environment, causing widespread pollution and posing risks to human health. However, despite their high environmental occurrence, there is very limited information regarding their toxicological effects. It is urgent to develop high-throughput screening (HTS) methods to guide toxicological studies. In this study, a receptor-ligand binding assay using an HTS system was developed to determine the binding potency of environmental pollutants on nuclear receptors. The test is conducted using a microplate reader (i.e., a 96-well plate containing various chemicals) by measuring the fluorescence polarization (FP) of a specific fluorescent probe. This assay consists of four parts: the construction and transformation of recombinant vectors, the expression and purification of the receptor protein (ligand-binding domain), receptor-probe binding, and competitive binding of chemicals with the receptor. The binding potency of two environmental pollutants, perfluorooctanesulfonic acid (PFOS) and triphenyl phosphate (TPHP), with peroxisome proliferator-activated receptor gamma (PPARγ) was determined to illustrate the assay procedure. Finally, the advantages and disadvantages of this method and its potential applications were also discussed.


Subject(s)
Environmental Pollutants , Fluorocarbons , High-Throughput Screening Assays , PPAR gamma , Receptors, Cytoplasmic and Nuclear , Environmental Pollutants/metabolism , Environmental Pollutants/chemistry , High-Throughput Screening Assays/methods , PPAR gamma/metabolism , PPAR gamma/chemistry , PPAR gamma/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/chemistry , Fluorocarbons/chemistry , Fluorocarbons/metabolism , Alkanesulfonic Acids/chemistry , Alkanesulfonic Acids/metabolism , Humans , Organophosphates/metabolism , Organophosphates/chemistry , Fluorescence Polarization/methods , Fluorescent Dyes/chemistry
7.
Mar Drugs ; 22(9)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39330276

ABSTRACT

Cyclophosphamide (CP) is an anticancer drug that causes infertility disorders. This study was designed to evaluate a nanoformulation of chitosan with an ethanolic extract from Spirulina platensis in terms of its protection against cyclophosphamide-induced ovarian toxicity. Nine groups of female Wistar rats were randomly assigned as follows: 1: control vehicle, 2: chitosan polymer, 3: telmisartan, 4: Spirulina platensis extract, 5: nanoformulation of the Spirulina platensis, and 6: single injection of CP; groups 7, 8, and 9 received the same treatments as those used in groups 3, 4, and 5, respectively, with a single dose of CP (200 mg/kg, I.P). The results displayed that the CP treatment decreased estradiol, progesterone, anti-mullerian hormone, and GSH content, and it downregulated PPAR-γ, Nrf-2, and HO-1 gene expression. In addition, the CP treatment caused an increase in the FSH, LH, and MDA levels. In the same manner, the protein expression of caspase-3, NF-kB, and TNF-α was upregulated in response to the CP treatment, while PPAR-γ was downregulated in comparison with the control. The rats treated with SPNPs exhibited a substantial reduction in the detrimental effects of oxidative stress and inflammation of the ovarian tissue. This study's conclusions showed that SPNPs counteracted the effects of CP, preventing the death of ovarian follicles and restoring the gonadotropin hormone balance and normal ovarian histological appearance.


Subject(s)
Chitosan , Cyclophosphamide , NF-E2-Related Factor 2 , NF-kappa B , Ovary , PPAR gamma , Tumor Necrosis Factor-alpha , Animals , Female , Rats , Chitosan/chemistry , Chitosan/pharmacology , Cyclophosphamide/toxicity , Ethanol/chemistry , Heme Oxygenase (Decyclizing)/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Ovary/drug effects , Ovary/pathology , Ovary/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Rats, Wistar , Signal Transduction/drug effects , Spirulina , Tumor Necrosis Factor-alpha/metabolism
8.
Medicine (Baltimore) ; 103(39): e39867, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331858

ABSTRACT

Sepsis, a multifaceted syndrome driven by an imbalanced host response to infection, remains a significant medical challenge. At its core lies the pivotal role of glycolysis, orchestrating immune responses especially in severe sepsis. The intertwined dynamics between glycolysis, sepsis, and immunity, however, have gaps in knowledge with several Crucial genes still shrouded in ambiguity. We harvested transcriptomic profiles from the peripheral blood of 107 septic patients juxtaposed against 29 healthy controls. Delving into this dataset, differential expression analysis shed light on genes distinctly linked to glycolysis in both cohorts. Harnessing the prowess of LASSO regression and SVM-RFE, we isolated Crucial genes, paving the way for a sepsis risk prediction model, subsequently vetted via Calibration and decision curve analysis. Using the CIBERSORT algorithm, we further mapped 22 immune cell subtypes within the septic samples, establishing potential interactions with the delineated Crucial genes. Our efforts unveiled 21 genes intricately tied to glycolysis that exhibited differential expression patterns. Gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses offered insights, spotlighting pathways predominantly associated with oxidative phosphorylation, PPAR signaling pathway, Glycolysis/Gluconeogenesis and HIF-1 signaling pathway. Among the myriad genes, IER3, DSC2, and PPARG emerged as linchpins, their prominence in sepsis further validated through ROC analytics. These sentinel genes demonstrated profound affiliations with various immune cell facets, bridging the complex terrain of glycolysis, sepsis, and immune responses. In line with our endeavor to "unveil the glycolysis in sepsis," the discovery of IER3, DSC2, and PPARG reinforces their cardinal roles in sepsis pathogenesis. These revelations accentuate the intricate dance between glycolysis and immunological shifts in septic conditions, offering novel avenues for therapeutic interventions.


Subject(s)
Computational Biology , Glycolysis , Machine Learning , PPAR gamma , Sepsis , Humans , Sepsis/genetics , Sepsis/immunology , Sepsis/metabolism , Glycolysis/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Computational Biology/methods , Male , Female , Middle Aged , Transcriptome
9.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337708

ABSTRACT

This study investigated the effects of ascochlorin (ASC), a natural compound derived from the fungus Ascochyta viciae, on adipogenesis and obesity. We determined the effects of ASC on 3T3-L1 preadipocytes and whether it ameliorated to mitigate high-fat diet (HFD)-induced obesity in C57BL/6J mice. We found that ASC significantly inhibited the differentiation of preadipocytes by modulating the Wnt/ß-catenin signaling pathway, a key regulator of adipogenic processes. Treatment with ASC not only reduced the mRNA and protein expression of key adipogenic transcription factors such as C/EBPα and PPARγ, but also reduced lipid accumulation both in vitro and in vivo. In addition, treatment HFD-fed mice with ASC significantly reduced their weight gain and adiposity vs. control mice. These results suggest that ASC has considerable potential as a therapeutic agent for obesity, owing to its dual action of inhibiting adipocyte differentiation and reducing lipid accumulation. Thus, ASC represents a promising candidate as a natural anti-obesity agent.


Subject(s)
Adipocytes , Adipogenesis , Diet, High-Fat , Obesity , Wnt Signaling Pathway , Animals , Male , Mice , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Alkenes , Anti-Obesity Agents/pharmacology , beta Catenin/metabolism , Cell Differentiation/drug effects , Diet, High-Fat/adverse effects , Lipid Metabolism/drug effects , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Obesity/etiology , Phenols/pharmacology , PPAR gamma/metabolism , PPAR gamma/genetics , Wnt Signaling Pathway/drug effects
10.
Nutrients ; 16(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39339767

ABSTRACT

BACKGROUND: Capsaicin, a bioactive compound found in peppers, is recognized for its anti-inflammatory, antioxidant, and anti-lipidemic properties. This study aimed to evaluate the effects of capsaicin on atherosclerosis progression. METHODS: Apolipoprotein E knockout mice and their C57BL/6 controls were utilized to assess blood lipid profile, inflammatory status, and atherosclerotic lesions. We also examined the influence of capsaicin on cholesterol influx and efflux, and the role of TRPV1 and PPARγ signaling pathways in bone marrow-derived macrophages. RESULTS: Capsaicin treatment reduced weight gain, visceral adiposity, blood triglycerides, and total and non-HDL cholesterol. These improvements were associated with a reduction in atherosclerotic lesions in the aorta and carotid. Capsaicin also improved hepatic oxidative and inflammatory status. Systemic inflammation was also reduced, as indicated by reduced leukocyte rolling and adhesion on the mesenteric plexus. Capsaicin decreased foam cell formation by reducing cholesterol influx through scavenger receptor A and increasing cholesterol efflux via ATP-binding cassette transporter A1, an effect primarily linked to TRPV1 activation. CONCLUSIONS: These findings underscore the potential of capsaicin as a promising agent for atherosclerosis prevention, highlighting its comprehensive role in modulating lipid metabolism, foam cell formation, and inflammatory responses.


Subject(s)
Atherosclerosis , Capsaicin , Foam Cells , Inflammation , PPAR gamma , TRPV Cation Channels , Animals , Male , Mice , Anti-Inflammatory Agents/pharmacology , Atherosclerosis/prevention & control , Atherosclerosis/drug therapy , ATP Binding Cassette Transporter 1/metabolism , Capsaicin/pharmacology , Cholesterol/blood , Cholesterol/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Inflammation/drug therapy , Lipid Metabolism/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , PPAR gamma/metabolism , Signal Transduction/drug effects , TRPV Cation Channels/metabolism
11.
Int Heart J ; 65(5): 929-938, 2024.
Article in English | MEDLINE | ID: mdl-39343596

ABSTRACT

Pulmonary arterial hypertension is a progressive heart and lung disease that is caused by irreversible pulmonary vascular remodeling. Sinomenine hydrochloride is an alkaloid that is extracted from sinomenium acutum, which has strong anti-inflammatory effects. In this study, male rats were injected with monocrotaline, and endothelial cells were exposed to hypoxia for 24 hours to induce pulmonary arterial hypertension. Apoptosis, inflammation, and oxidative stress pathways were observed the in lungs and cells. Sinomenine hydrochloride repressed the increased right ventricular systolic pressure and attenuated the right ventricular hypertrophy and pulmonary artery remodeling in model rats. It reversed the expression of BCL2 and BAX and prevented the apoptosis of endothelial cells. Additionally, it increased the contents of IKBα and NRF2. P65, P-P65, TNFα, IL1ß, and IL6 levels in the lungs decreased by it. Malondialdehyde contents decreased, and the superoxide dismutase and glutathione peroxidase activity and HO-1 level increased in the treatment group. In vivo, it promoted apoptosis of pulmonary artery endothelial cells. Moreover, by activating PPAR-γ, sinomenine hydrochloride attains the above effects. These data suggested that sinomenine hydrochloride could protect endothelial cells, restrain inflammation and oxidative stress, and enhance pulmonary vascular remodeling.


Subject(s)
Apoptosis , Endothelial Cells , Hypertension, Pulmonary , Morphinans , Oxidative Stress , PPAR gamma , Morphinans/pharmacology , Morphinans/therapeutic use , Animals , Apoptosis/drug effects , Male , Rats , PPAR gamma/metabolism , Oxidative Stress/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Vascular Remodeling/drug effects , Cells, Cultured
12.
PLoS One ; 19(9): e0310947, 2024.
Article in English | MEDLINE | ID: mdl-39325731

ABSTRACT

BACKGROUND: Kidney stone formation is a common disease that causes a significant threat to human health. The crystallization mechanism of calcium oxalate, the most common type of kidney stone, has been extensively researched, yet the damaging effects and mechanisms of calcium oxalate crystals on renal tubular epithelial cells remain incompletely elucidated. Regulated mitochondrial dynamics is essential for eukaryotic cells, but its role in the occurrence and progression of calcium oxalate (CaOx) nephrolithiasis is not yet understood. METHODS: An animal model of calcium oxalate-related nephrolithiasis was established in adult male Sprague‒Dawley (SD) rats by continuously administering drinking water containing 1% ethylene glycol for 28 days. The impact of calcium oxalate crystals on mitochondrial dynamics and apoptosis in renal tubular epithelial cells was investigated using HK2 cells in vitro. Blood samples and bilateral kidney tissues were collected for histopathological evaluation and processed for tissue injury, inflammation, fibrosis, oxidative stress detection, and mitochondrial dynamics parameter analysis. RESULTS: Calcium oxalate crystals caused higher levels of mitochondrial fission and apoptosis in renal tubular epithelial cells both in vivo and in vitro. Administration of a PPARγ agonist significantly alleviated mitochondrial fission and apoptosis in renal tubular epithelial cells, and improved renal function, accompanied by reduced levels of oxidative stress, increased antioxidant enzyme expression, alleviation of inflammation, and reduced fibrosis in vivo. CONCLUSION: Our results indicated that increased mitochondrial fission in renal tubular epithelial cells is a critical component of kidney injury caused by calcium oxalate stones, leading to the accumulation of reactive oxygen species within the tissue and the subsequent initiation of apoptosis. Regulating mitochondrial dynamics represents a promising approach for calcium oxalate nephrolithiasis.


Subject(s)
Apoptosis , Calcium Oxalate , Epithelial Cells , Kidney Tubules , Mitochondrial Dynamics , Nephrolithiasis , PPAR gamma , Rats, Sprague-Dawley , Animals , Male , Mitochondrial Dynamics/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Nephrolithiasis/metabolism , Nephrolithiasis/drug therapy , Nephrolithiasis/pathology , Rats , PPAR gamma/metabolism , PPAR gamma/agonists , Calcium Oxalate/metabolism , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Apoptosis/drug effects , Humans , Cell Line , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Disease Models, Animal
13.
Respir Res ; 25(1): 345, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39313791

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung epithelial phenotypes, fibroblast activation, and increased extracellular matrix deposition. Transforming growth factor-beta (TGF-ß)1-induced Smad signaling and downregulation of peroxisomal genes are involved in the pathogenesis and can be inhibited by peroxisome proliferator-activated receptor (PPAR)-α activation. However, the three PPARs, that is PPAR-α, PPAR-ß/δ, and PPAR-γ, are known to interact in a complex crosstalk. METHODS: To mimic the pathogenesis of lung fibrosis, primary lung fibroblasts from control and IPF patients with comparable levels of all three PPARs were treated with TGF-ß1 for 24 h, followed by the addition of PPAR ligands either alone or in combination for another 24 h. Fibrosis markers (intra- and extracellular collagen levels, expression and activity of matrix metalloproteinases) and peroxisomal biogenesis and metabolism (gene expression of peroxisomal biogenesis and matrix proteins, protein levels of PEX13 and catalase, targeted and untargeted lipidomic profiles) were analyzed after TGF-ß1 treatment and the effects of the PPAR ligands were investigated. RESULTS: TGF-ß1 induced the expected phenotype; e.g. it increased the intra- and extracellular collagen levels and decreased peroxisomal biogenesis and metabolism. Agonists of different PPARs reversed TGF-ß1-induced fibrosis even when given 24 h after TGF-ß1. The effects included the reversals of (1) the increase in collagen production by repressing COL1A2 promoter activity (through PPAR-ß/δ activation); (2) the reduced activity of matrix metalloproteinases (through PPAR-ß/δ activation); (3) the decrease in peroxisomal biogenesis and lipid metabolism (through PPAR-γ activation); and (4) the decrease in catalase protein levels in control (through PPAR-γ activation) and IPF (through a combined activation of PPAR-ß/δ and PPAR-γ) fibroblasts. Further experiments to explore the role of catalase showed that an overexpression of catalase protein reduced collagen production. Additionally, the beneficial effect of PPAR-γ but not of PPAR-ß/δ activation on collagen synthesis depended on catalase activity and was thus redox-sensitive. CONCLUSION: Our data provide evidence that IPF patients may benefit from a combined activation of PPAR-ß/δ and PPAR-γ.


Subject(s)
Idiopathic Pulmonary Fibrosis , PPAR delta , PPAR gamma , PPAR-beta , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , PPAR-beta/metabolism , PPAR-beta/genetics , PPAR-beta/agonists , Cells, Cultured , PPAR delta/metabolism , PPAR delta/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/drug effects , Peroxisomes/metabolism , Peroxisomes/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Male , Transforming Growth Factor beta1/metabolism , Female
14.
Int J Oncol ; 65(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39301639

ABSTRACT

Macrophages have crucial roles in immune responses and tumor progression, exhibiting diverse phenotypes based on environmental cues. In the present study, the impact of cinobufagin (CB) on macrophage polarization and the consequences on tumor­associated behaviors were investigated. Morphological transformations of THP­1 cells into M0, M1 and M2 macrophages were observed, including distinct changes in the size, shape and adherence properties of these cells. CB treatment inhibited the viability of A549 and LLC cells in a concentration­dependent manner, with an IC50 of 28.8 and 30.12 ng/ml, respectively. CB at concentrations of <30 ng/ml had no impact on the viability of M0 macrophages and lung epithelial (BEAS­2B) cells. CB influenced the expression of macrophage surface markers, reducing CD206 positivity in M2 macrophages without affecting CD86 expression in M1 macrophages. CB also altered certain expression profiles at the mRNA level, notably downregulating macrophage receptor with collagenous structure (MARCO) expression in M2 macrophages and upregulating tumor necrosis factor­α and interleukin­1ß in both M0 and M1 macrophages. Furthermore, ELISA analyses revealed that CB increased the levels of pro­inflammatory cytokines in M1 macrophages and reduced the levels of anti­inflammatory factors in M2 macrophages. CB treatment also attenuated the migration and invasion capacities of A549 and LLC cells stimulated by M2 macrophage­conditioned medium. Additionally, CB modulated peroxisome proliferator­activated receptor γ (PPARγ) and MARCO expression in M2 macrophages and epithelial­mesenchymal transition in A549 cells, which was partially reversed by rosiglitazone, a PPARγ agonist. Finally, CB and cisplatin treatments hindered tumor growth in vivo, with distinct impacts on animal body weight and macrophage marker expression in tumor tissues. In conclusion, the results of the present study demonstrated that CB exerted complex regulatory effects on macrophage polarization and tumor progression, suggesting its potential as a modulator of the tumor microenvironment and a therapeutic for cancer treatment.


Subject(s)
Bufanolides , Cell Movement , Lung Neoplasms , Neoplasm Invasiveness , Tumor-Associated Macrophages , Bufanolides/pharmacology , Bufanolides/therapeutic use , Humans , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/immunology , Animals , Mice , Cell Movement/drug effects , A549 Cells , Xenograft Model Antitumor Assays , THP-1 Cells , PPAR gamma/metabolism , Macrophage Activation/drug effects , Cell Line, Tumor
15.
Clin Exp Pharmacol Physiol ; 51(11): e13917, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39285148

ABSTRACT

Ischaemic stroke is a common condition that can lead to cerebral ischaemia-reperfusion injury. Phillygenin (PHI), a natural bioactive compound derived from Forsythia suspensa, has been shown to play a crucial role in regulating inflammation across various diseases. However, its specific regulatory effects in ischaemic stroke progression remain unclear. In this study, we established a middle cerebral artery occlusion (MCAO) rat model. Treatment with PHI (50 or 100 mg/kg) significantly reduced cerebral infarction in MCAO rats. PHI treatment also mitigated the increased inflammatory response observed in these rats. Additionally, PHI suppressed microglial activation by reducing iNOS expression, a marker of M1-type polarization of microglia, and attenuated increased brain tissue apoptosis in MCAO rats. Furthermore, PHI's anti-inflammatory effects in MCAO rats were abrogated upon co-administration with GW9662, a peroxisome proliferator-activated receptor γ (PPARγ) inhibitor. In summary, PHI attenuated microglial activation and apoptosis in cerebral ischaemia-reperfusion injury through PPARγ activation, suggesting its potential as a therapeutic agent for mitigating cerebral ischaemia-reperfusion injury.


Subject(s)
Apoptosis , Infarction, Middle Cerebral Artery , Microglia , PPAR gamma , Rats, Sprague-Dawley , Reperfusion Injury , Animals , PPAR gamma/metabolism , Apoptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Male , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Lignans
16.
Chem Res Toxicol ; 37(9): 1574-1587, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39235066

ABSTRACT

ZLY06 is a dual agonist of peroxisome proliferator-activated receptor (PPAR) δ/γ, showing potential therapeutic effects on metabolic syndrome. However, our research has revealed that ZLY06 exhibits hepatotoxicity in normal C57BL/6J mice, though the precise mechanism remains unclear. This study aims to investigate the manifestations and mechanisms of ZLY06-induced hepatotoxicity. We administered ZLY06 via oral gavage to C57BL/6J mice (once daily for six weeks) and monitored various indicators to preliminarily explore its hepatotoxicity. Additionally, we further investigate the specific mechanisms of ZLY06-induced hepatotoxicity using PPAR inhibitors (GW9662 and GSK0660) and the Protein kinase B (AKT) activator (SC79). Results showed that ZLY06 led to increased serum ALP, ALT and AST, as well as elevated liver index and hepatic lipid levels. There was upregulation in the gene and protein expression of lipid metabolism-related molecules Acc, Scd1, Cd36, Fabp1 and Fabp2 in hepatocytes, with Cd36 showing the most significant change. Furthermore, cotreatment with SC79 significantly reduced ZLY06-induced hepatotoxicity in AML12 cells, evidenced by decreased intracellular TG levels and downregulation of CD36 expression. Specific knockdown of CD36 also mitigated ZLY06-induced hepatotoxicity. The study found that ZLY06 may bind to AKT1, inhibiting its phosphorylation activation, with the downregulation of p-AKT1 preceding the upregulation of CD36. In summary, ZLY06 mediates the upregulation of CD36 by potentially binding to and inhibiting the phosphorylation of AKT1, leading to hepatic lipid metabolism disorder and inducing liver toxicity.


Subject(s)
CD36 Antigens , Lipid Metabolism , Liver , Mice, Inbred C57BL , PPAR gamma , Proto-Oncogene Proteins c-akt , Up-Regulation , Animals , CD36 Antigens/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation/drug effects , Mice , Up-Regulation/drug effects , Liver/drug effects , Liver/metabolism , Male , PPAR gamma/agonists , PPAR gamma/metabolism , Lipid Metabolism/drug effects , PPAR delta/metabolism , PPAR delta/agonists , PPAR delta/antagonists & inhibitors
17.
Ecotoxicol Environ Saf ; 284: 116988, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39236653

ABSTRACT

Hypospadias is one of the most common congenital anomalies of the male urogenital system, and di(2-ethylhexyl) phthalate (DEHP), a widely used endocrine-disrupting chemical (EDC), is considered a significant risk factor for this condition. Mono-2-ethylhexyl phthalate (MEHP), the toxic active metabolite of DEHP, has been proven to affect penile development and ultimately result in the hypospadias phenotype. However, while it is acknowledged that hypospadias arises from the aberrant development of multiple penile tissues, the specific impact of MEHP on human foreskin tissue development and its underlying molecular mechanisms of action remain unclear. In this study, we constructed an in vitro toxicity assay for MEHP using human foreskin fibroblasts and employed high-throughput RNA sequencing to investigate the molecular mechanisms subserving the defects in cellular function. We subsequently conducted multi-omics data analysis using public databases to analyze key target genes, and identified MMP11 as a chief downstream gene responsible for the effects of MEHP on HFF-1 cell migration. Through molecular docking analysis and molecular biology experiments, we further demonstrated that the nuclear receptor PPAR-gamma was activated upon binding with MEHP, leading to the suppression of MMP11 expression. Additionally, we found that epigenetic modifications induced by MEHP were also involved in its pathogenic effects on hypospadias. Our research highlights the crucial role of impaired cellular proliferation and migration in MEHP-induced hypospadias. We identified the MEHP/PPAR-gamma/MMP11 pathway as a novel pathogenic mechanism, providing important potential targets for future preventive strategies with respect to hypospadias.


Subject(s)
Diethylhexyl Phthalate , Fibroblasts , Foreskin , Hypospadias , Matrix Metalloproteinase 11 , Humans , Male , Cell Movement/drug effects , Diethylhexyl Phthalate/toxicity , Diethylhexyl Phthalate/analogs & derivatives , Down-Regulation/drug effects , Endocrine Disruptors/toxicity , Fibroblasts/drug effects , Foreskin/cytology , Foreskin/metabolism , Hypospadias/chemically induced , Hypospadias/pathology , Matrix Metalloproteinase 11/genetics , Molecular Docking Simulation , PPAR gamma/metabolism , PPAR gamma/genetics
18.
Biochem Biophys Res Commun ; 733: 150690, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39276693

ABSTRACT

The prevalence of obesity and its associated metabolic disorders has emerged as one of the most significant health threats worldwide. The visceral adipose tissue regulatory T cells (VAT Treg) play an essential role in maintaining homeostasis and preventing obesity mainly by secreting Interleikin-10 (IL-10) and Transforming Growth Factor ß (TGF-ß). However, the mechanism that regulates VAT Treg quantity and function remains unclear. Here we elucidate the pivotal role of IL-27 signaling in sustaining the accumulation of VAT Treg cells, thereby conferring protection against obesity. We found that mice with the deficiency of IL-27 receptor Wsx1 gained more body weight and VAT weight than their wild-type littermates when fed both a normal-fat diet (NFD) and a high-fat diet (HFD). Notably, the population of VAT Treg cells was reduced in Wsx1 knockout (KO) mice, regardless of whether they were fed a normal-fat diet (NFD) or a high-fat diet (HFD). Correspondingly, the expression levels of the transcription factors FOXP3 and PPAR-γ, essential for VAT Treg function, were also diminished in Wsx1 KO mice. Taken together, our findings indicate that IL-27 signaling plays a protective role in obesity by supporting the maintenance and accumulation of VAT Treg cells.


Subject(s)
Diet, High-Fat , Intra-Abdominal Fat , Mice, Inbred C57BL , Mice, Knockout , Obesity , Receptors, Interleukin , Signal Transduction , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Obesity/metabolism , Obesity/immunology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/immunology , Receptors, Interleukin/metabolism , Receptors, Interleukin/genetics , Diet, High-Fat/adverse effects , Mice , Male , PPAR gamma/metabolism , Interleukins/metabolism , Interleukins/genetics
19.
J Biochem Mol Toxicol ; 38(10): e23855, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39328005

ABSTRACT

Adriamycin is an anticancer anthracycline drug that inhibits the progression of topoisomerase II activity and causes apoptosis. The effective clinical application of the drug is very much limited by its adverse drug reactions on various tissues. Most importantly, Adriamycin causes cardiomyopathy, one of the life-threatening complications of the drug. Altered expression of PPARγ in adipocytes inhibited the glucose and fatty acids uptake by down regulating GLUT4 and CD36 expression and causes cardiotoxicity. Therefore, the influence of Adriamycin in cardiac ailments was investigated in vivo and in vitro. Adriamycin treated rats showed altered ECG profile, arrhythmic heartbeat with the elevated levels of CRP and LDH. Dysregulated lipid profiles with elevated levels of cholesterol and triglycerides were also observed. Possibilities of cardiac problems due to cardiomyopathy were analyzed through histopathology. Adriamycin treated rats showed no signs for atheromatous plaque formation in aorta but disorganized cardiomyocytes with myofibrillar loss and inflammation in heart tissue, indicative of cardiomyopathy. Reduced levels of antioxidant enzymes confirmed the incidence of oxidative stress. Adriamycin treatment significantly reduced glucose and insulin levels, creating energy demand due to decreased glucose and insulin levels with increased fatty acid accumulation, ultimately resulting in oxidative stress mediated cardiomyopathy. Since PPARs play a vital role in regulating oxidative stress, the effect of Adriamycin on PPARγ was analyzed by western blot. Adriamycin downregulated PPARγ in a dose-dependent manner in H9C2 cells in vitro. Overall, our study suggests that Adriamycin alters glucose and lipid metabolism via PPARγ inhibition that leads to oxidative stress and cardiomyopathy that necessitates a different therapeutic approach.


Subject(s)
Cardiomyopathies , Doxorubicin , PPAR gamma , Animals , Male , Rats , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Line , Doxorubicin/adverse effects , Energy Metabolism/drug effects , Oxidative Stress/drug effects , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Rats, Wistar
20.
Nutrients ; 16(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339665

ABSTRACT

Obesity and type 2 diabetes mellitus are global public health issues. Although males show higher obesity and insulin resistance prevalence, current treatments often neglect sex-specific differences. White adipose tissue (WAT) is crucial in preventing lipotoxicity and inflammation and has become a key therapeutic target. Rosiglitazone (RSG), a potent PPARγ agonist, promotes healthy WAT growth and mitochondrial function through MitoNEET modulation. Recent RSG-based strategies specifically target white adipocytes, avoiding side effects. Our aim was to investigate whether sex-specific differences in the insulin-sensitizing effects of RSG exist on WAT during obesity and inflammation. We used Wistar rats of both sexes fed a high-fat diet (HFD, 22.5% fat content) for 16 weeks. Two weeks before sacrifice, a group of HFD-fed rats received RSG treatment (4 mg/kg of body weight per day) within the diet. HFD male rats showed greater insulin resistance, inflammation, mitochondrial dysfunction, and dyslipidemia than females. RSG had more pronounced effects in males, significantly improving insulin sensitivity, fat storage, mitochondrial function, and lipid handling in WAT while reducing ectopic fat deposition and enhancing adiponectin signaling in the liver. Our study suggests a significant sexual dimorphism in the anti-diabetic effects of RSG on WAT, correlating with the severity of metabolic dysfunction.


Subject(s)
Adipose Tissue, White , Diet, High-Fat , Insulin Resistance , Rosiglitazone , Animals , Female , Male , Rats , Adiponectin/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Inflammation/drug therapy , Liver/metabolism , Liver/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/drug therapy , Obesity/metabolism , PPAR gamma/metabolism , PPAR gamma/agonists , Rats, Wistar , Rosiglitazone/pharmacology , Sex Characteristics , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL